Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 116
Filtrar
1.
BMJ Glob Health ; 8(8)2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37652566

RESUMO

New vector-control technologies to fight mosquito-borne diseases are urgently needed, the adoption of which depends on efficacy estimates from large-scale cluster-randomised trials (CRTs). The release of Wolbachia-infected mosquitoes is one promising strategy to curb dengue virus (DENV) transmission, and a recent CRT reported impressive reductions in dengue incidence following the release of these mosquitoes. Such trials can be affected by multiple sources of bias, however. We used mathematical models of DENV transmission during a CRT of Wolbachia-infected mosquitoes to explore three such biases: human movement, mosquito movement and coupled transmission dynamics between trial arms. We show that failure to account for each of these biases would lead to underestimated efficacy, and that the majority of this underestimation is due to a heretofore unrecognised bias caused by transmission coupling. Taken together, our findings suggest that Wolbachia-infected mosquitoes could be even more promising than the recent CRT suggested. By emphasising the importance of accounting for transmission coupling between arms, which requires a mathematical model, we highlight the key role that models can play in interpreting and extrapolating the results from trials of vector control interventions.


Assuntos
Doenças Transmitidas por Vetores , Animais , Humanos , Doenças Transmitidas por Vetores/prevenção & controle , Doenças Transmitidas por Vetores/transmissão , Culicidae , Viés , Modelos Biológicos
2.
Science ; 377(6614): eabc2757, 2022 09 30.
Artigo em Inglês | MEDLINE | ID: mdl-36173836

RESUMO

Many endemic poverty-associated diseases, such as malaria and leishmaniasis, are transmitted by arthropod vectors. Pathogens must interact with specific molecules in the vector gut, the microbiota, and the vector immune system to survive and be transmitted. The vertebrate host, in turn, is infected when the pathogen and vector-derived factors, such as salivary proteins, are delivered into the skin by a vector bite. Here, we review recent progress in our understanding of the biology of pathogen transmission from the human to the vector and back, from the vector to the host. We also highlight recent advances in the biology of vector-borne disease transmission, which have translated into additional strategies to prevent human disease by either reducing vector populations or by disrupting their ability to transmit pathogens.


Assuntos
Vetores Artrópodes , Interações Hospedeiro-Patógeno , Proteínas e Peptídeos Salivares , Doenças Transmitidas por Vetores , Animais , Vetores Artrópodes/microbiologia , Vetores Artrópodes/parasitologia , Humanos , Leishmaniose/prevenção & controle , Leishmaniose/transmissão , Malária/prevenção & controle , Malária/transmissão , Proteínas e Peptídeos Salivares/metabolismo , Doenças Transmitidas por Vetores/prevenção & controle , Doenças Transmitidas por Vetores/transmissão
3.
PLos ONE ; 17(9): 1-25, set. 2022. mapas, graf, ilus
Artigo em Inglês | RDSM | ID: biblio-1531381

RESUMO

Characterizing persistent malaria transmission that occurs after the combined deployment of indoor residual spraying (IRS) and long-lasting insecticidal nets (LLINs) is critical to guide malaria control and elimination efforts. This requires a detailed understanding of both human and vector behaviors at the same temporal and spatial scale. Cross-sectional human behavior evaluations and mosquito collections were performed in parallel in Magude district, Mozambique. Net use and the exact time when participant moved into each of five environments (outdoor, indoor before bed, indoor in bed, indoor after getting up, and outdoor after getting up) were recorded for individuals from three different age groups and both sexes during a dry and a rainy season. Malaria mosquitoes were collected with CDC light traps in combination with collection bottle rotators. The percentage of residual exposure to host-seeking vectors that occurred in each environment was calculated for five local malaria vectors with different biting behaviors, and the actual (at observed levels of LLIN use) and potential (i.e. if all residents had used an LLIN) personal protection conferred by LLINs was estimated. Anopheles arabiensis was responsible for more than 74% of residents' residual exposure to host-seeking vectors during the Magude project. The other four vector species (An. funestus s.s., An. parensis, An. squamosus and An. merus) were responsible for less than 10% each. The personal protection conferred by LLINs prevented only 39.2% of the exposure to host-seeking vectors that survived the implementation of both IRS and LLINs, and it differed significantly across seasons, vector species and age groups. At the observed levels of bednet use, 12.5% of all residual exposure to host-seeking vectors occurred outdoor during the evening, 21.9% indoor before going to bed, almost two thirds (64%) while people were in bed, 1.4% indoors after getting up and 0.2% outdoor after leaving the house. Almost a third of the residual exposure to host-seeking vectors (32.4%) occurred during the low transmission season. The residual bites of An. funestus s.s. and An. parensis outdoors and indoor before bedtime, of An. arabiensis indoors when people are in bed, and of An. squamosus both indoors and outdoors, are likely to have sustained malaria transmission throughout the Magude project. By increasing LLIN use, an additional 24.1% of exposure to the remaining hosts-seeking vectors could have been prevented. Since An. arabiensis, the most abundant vector, feeds primarily while people are in bed, increasing net use and net feeding inhibition (through e.g. community awareness activities and the selection of more effective LLINs) could significantly reduce the exposure to remaining host-seeking mosquitoes. Nonetheless, supplementary interventions aiming to reduce human-vector contact outdoors and/or indoors before people go to bed (e.g. through larval source management, window and eave screening, eave tubes, and spatial repellents) will be needed to reduce residual exposure to the outdoor and early biting An. funestus s.s. and An. parensis.


Assuntos
Humanos , Animais , Masculino , Feminino , Doenças Transmitidas por Vetores/transmissão , Inseticidas , Malária/prevenção & controle , Anopheles/fisiologia , Controle Biológico de Vetores/tendências , Controle de Mosquitos/estatística & dados numéricos , Estudos Transversais , Receptores Proteína Tirosina Quinases , Receptores Proteína Tirosina Quinases/imunologia , Progressão da Doença , Mosquitos Vetores , Moçambique
4.
Proc Natl Acad Sci U S A ; 119(26): e2118283119, 2022 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-35737833

RESUMO

Over half the world's population is at risk for viruses transmitted by Aedes mosquitoes, such as dengue and Zika. The primary vector, Aedes aegypti, thrives in urban environments. Despite decades of effort, cases and geographic range of Aedes-borne viruses (ABVs) continue to expand. Rigorously proven vector control interventions that measure protective efficacy against ABV diseases are limited to Wolbachia in a single trial in Indonesia and do not include any chemical intervention. Spatial repellents, a new option for efficient deployment, are designed to decrease human exposure to ABVs by releasing active ingredients into the air that disrupt mosquito-human contact. A parallel, cluster-randomized controlled trial was conducted in Iquitos, Peru, to quantify the impact of a transfluthrin-based spatial repellent on human ABV infection. From 2,907 households across 26 clusters (13 per arm), 1,578 participants were assessed for seroconversion (primary endpoint) by survival analysis. Incidence of acute disease was calculated among 16,683 participants (secondary endpoint). Adult mosquito collections were conducted to compare Ae. aegypti abundance, blood-fed rate, and parity status through mixed-effect difference-in-difference analyses. The spatial repellent significantly reduced ABV infection by 34.1% (one-sided 95% CI lower limit, 6.9%; one-sided P value = 0.0236, z = 1.98). Aedes aegypti abundance and blood-fed rates were significantly reduced by 28.6 (95% CI 24.1%, ∞); z = -9.11) and 12.4% (95% CI 4.2%, ∞); z = -2.43), respectively. Our trial provides conclusive statistical evidence from an appropriately powered, preplanned cluster-randomized controlled clinical trial of the impact of a chemical intervention, in this case a spatial repellent, to reduce the risk of ABV transmission compared to a placebo.


Assuntos
Aedes , Repelentes de Insetos , Controle de Mosquitos , Mosquitos Vetores , Doenças Transmitidas por Vetores , Adulto , Animais , Dengue/epidemiologia , Dengue/prevenção & controle , Humanos , Controle de Mosquitos/normas , Peru/epidemiologia , Doenças Transmitidas por Vetores/epidemiologia , Doenças Transmitidas por Vetores/prevenção & controle , Doenças Transmitidas por Vetores/transmissão , Zika virus , Infecção por Zika virus
5.
Sci Rep ; 12(1): 2066, 2022 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-35136077

RESUMO

In this study, we utilized an untargeted NMR metabolomics approach to identify the vector response in terms of metabolic profiling after temperature and insecticide exposure in comparison with the control. Clearly, temperature and insecticide exposure cause changes in the underlying metabolism, and the NMR metabolomic profile enables a direct examination of the immediate response of the vector to cope up with these changes. The present study was designed in four parts: A-Aedes aegypti were exposed to 40 °C for one-hour, DDT-4%, malathion-5%, and deltamethrin-0.05% separately and, part B-D; one-hour exposure at 35 °C and 40 °C temperatures followed by one-hour exposure to insecticide. The resultant metabolite profiles were compared with the control. In response to temperature and insecticide exposure, several metabolites and altered pathways were identified. Citrate, maltose, lipids, Nicotinate, Choline, Pyruvate and ß-hydroxybutyrate were found as important components of major biological pathways such as tri-carboxylic acid cycle, branched amino acid degradation, glycolysis/gluconeogenesis, amino acid metabolism, lipid and carbohydrate metabolism, nucleotide PRPP pathway, and phospholipid metabolism. Furthermore, the results also suggest that the changes imposed by exposure to temperature and insecticides individually, are reversed with combined exposure, thus negating the impact of each other and posing a threat to the control of Aedes-borne diseases such as dengue, chikungunya, Zika and yellow fever.


Assuntos
Aedes/metabolismo , Inseticidas/farmacologia , Redes e Vias Metabólicas/efeitos dos fármacos , Metaboloma/efeitos dos fármacos , Doenças Transmitidas por Vetores/transmissão , Aedes/efeitos dos fármacos , Animais , DDT/farmacologia , Resistência a Inseticidas/fisiologia , Malation/farmacologia , Metabolômica/métodos , Mosquitos Vetores/efeitos dos fármacos , Nitrilas/farmacologia , Piretrinas/farmacologia , Temperatura
6.
PLoS Negl Trop Dis ; 16(2): e0010186, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35176020

RESUMO

BACKGROUND: In Greece vector borne diseases (VBD) and foremost West Nile virus (WNV) pose an important threat to public health and the tourist industry, the primary sector of contribution to the national economy. The island of Crete, is one of Greece's major tourist destinations receiving annually over 5 million tourists making regional VBD control both a public health and economic priority. METHODOLOGY: Under the auspices of the Region of Crete, a systematic integrative surveillance network targeting mosquitoes and associated pathogens was established in Crete for the years 2018-2020. Using conventional and molecular diagnostic tools we investigated the mosquito species composition and population dynamics, pathogen infection occurrences in vector populations and in sentinel chickens, and the insecticide resistance status of the major vector species. PRINCIPAL FINDINGS: Important disease vectors were recorded across the island including Culex pipiens, Aedes albopictus, and Anopheles superpictus. Over 75% of the sampled specimens were collected in the western prefectures potentially attributed to the local precipitation patterns, with Cx. pipiens being the most dominant species. Although no pathogens (flaviviruses) were detected in the analysed mosquito specimens, chicken blood serum analyses recorded a 1.7% WNV antibody detection rate in the 2018 samples. Notably detection of the first WNV positive chicken preceded human WNV occurrence in the same region by approximately two weeks. The chitin synthase mutation I1043F (associated with high diflubenzuron resistance) was recorded at an 8% allelic frequency in Lasithi prefecture Cx. pipiens mosquitoes (sampled in 2020) for the first time in Greece. Markedly, Cx. pipiens populations in all four prefectures were found harboring the kdr mutations L1014F/C/S (associated with pyrethroid resistance) at a close to fixation rate, with mutation L1014C being the most commonly found allele (≥74% representation). Voltage gated sodium channel analyses in Ae. albopictus revealed the presence of the kdr mutations F1534C and I1532T (associated with putative mild pyrethroid resistance phenotypes) yet absence of V1016G. Allele F1534C was recorded in all prefectures (at an allelic frequency range of 25-46.6%) while I1532T was detected in populations from Chania, Rethymnon and Heraklion (at frequencies below 7.1%). Finally, no kdr mutations were detected in the Anopheles specimens included in the analyses. CONCLUSIONS/SIGNIFICANCE: The findings of our study are of major concern for VBD control in Crete, highlighting (i) the necessity for establishing seasonal integrated entomological/pathogen surveillance programs, supporting the design of targeted vector control responses and; ii) the need for establishing appropriate insecticide resistance management programs ensuring the efficacy and sustainable use of DFB and pyrethroid based products in vector control.


Assuntos
Culicidae/efeitos dos fármacos , Resistência a Inseticidas , Inseticidas/farmacologia , Mosquitos Vetores/efeitos dos fármacos , Doenças Transmitidas por Vetores/veterinária , Doenças Transmitidas por Vetores/virologia , Febre do Nilo Ocidental/veterinária , Febre do Nilo Ocidental/virologia , Animais , Galinhas , Culicidae/classificação , Culicidae/fisiologia , Culicidae/virologia , Diflubenzuron/farmacologia , Grécia , Humanos , Proteínas de Insetos/genética , Proteínas de Insetos/metabolismo , Mosquitos Vetores/classificação , Mosquitos Vetores/genética , Mosquitos Vetores/fisiologia , Mutação , Doenças das Aves Domésticas/transmissão , Doenças das Aves Domésticas/virologia , Piretrinas/farmacologia , Doenças Transmitidas por Vetores/transmissão , Febre do Nilo Ocidental/transmissão , Vírus do Nilo Ocidental/genética , Vírus do Nilo Ocidental/isolamento & purificação , Vírus do Nilo Ocidental/fisiologia
7.
Sci Rep ; 12(1): 1748, 2022 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-35110661

RESUMO

African horse sickness is a vector-borne, non-contagious and highly infectious disease of equines caused by African horse sickness viruses (AHSv) that mainly affect horses. The occurrence of the disease causes huge economic impacts because of its high fatality rate, trade ban and disease control costs. In the planning of vectors and vector-borne diseases like AHS, the application of Ecological niche models (ENM) used an enormous contribution in precisely delineating the suitable habitats of the vector. We developed an ENM to delineate the global suitability of AHSv based on retrospective outbreak data records from 2005 to 2019. The model was developed in an R software program using the Biomod2 package with an Ensemble modeling technique. Predictive environmental variables like mean diurnal range, mean precipitation of driest month(mm), precipitation seasonality (cv), mean annual maximum temperature (oc), mean annual minimum temperature (oc), mean precipitation of warmest quarter(mm), mean precipitation of coldest quarter (mm), mean annual precipitation (mm), solar radiation (kj /day), elevation/altitude (m), wind speed (m/s) were used to develop the model. From these variables, solar radiation, mean maximum temperature, average annual precipitation, altitude and precipitation seasonality contributed 36.83%, 17.1%, 14.34%, 7.61%, and 6.4%, respectively. The model depicted the sub-Sahara African continent as the most suitable area for the virus. Mainly Senegal, Burkina Faso, Niger, Nigeria, Ethiopia, Sudan, Somalia, South Africa, Zimbabwe, Madagascar and Malawi are African countries identified as highly suitable countries for the virus. Besides, OIE-listed disease-free countries like India, Australia, Brazil, Paraguay and Bolivia have been found suitable for the virus. This model can be used as an epidemiological tool in planning control and surveillance of diseases nationally or internationally.


Assuntos
Vírus da Doença Equina Africana , Doença Equina Africana , Ecossistema , Modelos Estatísticos , África/epidemiologia , Doença Equina Africana/epidemiologia , Doença Equina Africana/transmissão , Animais , Ceratopogonidae/virologia , Surtos de Doenças/veterinária , Cavalos , Índia/epidemiologia , Insetos Vetores/virologia , Software , África do Sul/epidemiologia , América do Sul/epidemiologia , Temperatura , Doenças Transmitidas por Vetores/epidemiologia , Doenças Transmitidas por Vetores/transmissão , Doenças Transmitidas por Vetores/veterinária
8.
Parasit Vectors ; 15(1): 37, 2022 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-35073983

RESUMO

BACKGROUND: The distribution of parasite load across hosts may modify the transmission dynamics of infectious diseases. Chagas disease is caused by a multi-host protozoan, Trypanosoma cruzi, but the association between host parasitemia and infectiousness to the vector has not been studied in sylvatic mammalian hosts. We quantified T. cruzi parasite load in sylvatic mammals, modeled the association of the parasite load with infectiousness to the vector and compared these results with previous ones for local domestic hosts. METHODS: The bloodstream parasite load in each of 28 naturally infected sylvatic mammals from six species captured in northern Argentina was assessed by quantitative PCR, and its association with infectiousness to the triatomine Triatoma infestans was evaluated, as determined by natural or artificial xenodiagnosis. These results were compared with our previous results for 88 humans, 70 dogs and 13 cats, and the degree of parasite over-dispersion was quantified and non-linear models fitted to data on host infectiousness and bloodstream parasite load. RESULTS: The parasite loads of Didelphis albiventris (white-eared opossum) and Dasypus novemcinctus (nine-banded armadillo) were directly and significantly associated with infectiousness of the host and were up to 190-fold higher than those in domestic hosts. Parasite load was aggregated across host species, as measured by the negative binomial parameter, k, and found to be substantially higher in white-eared opossums, cats, dogs and nine-banded armadillos (range: k = 0.3-0.5) than in humans (k = 5.1). The distribution of bloodstream parasite load closely followed the "80-20 rule" in every host species examined. However, the 20% of human hosts, domestic mammals or sylvatic mammals exhibiting the highest parasite load accounted for 49, 25 and 33% of the infected triatomines, respectively. CONCLUSIONS: Our results support the use of bloodstream parasite load as a proxy of reservoir host competence and individual transmissibility. The over-dispersed distribution of T. cruzi bloodstream load implies the existence of a fraction of highly infectious hosts that could be targeted to improve vector-borne transmission control efforts toward interruption transmission. Combined strategies that decrease the parasitemia and/or host-vector contact with these hosts would disproportionally contribute to T. cruzi transmission control.


Assuntos
Doença de Chagas/transmissão , Mamíferos/parasitologia , Triatoma/parasitologia , Trypanosoma cruzi , Animais , Animais Selvagens/parasitologia , Argentina/epidemiologia , Tatus/parasitologia , Gatos , Doença de Chagas/diagnóstico , Doença de Chagas/prevenção & controle , Didelphis/parasitologia , Reservatórios de Doenças/parasitologia , Vetores de Doenças , Cães , Florestas , Genes de Protozoários , Humanos , Insetos Vetores/parasitologia , Carga Parasitária/estatística & dados numéricos , Parasitemia/parasitologia , Reação em Cadeia da Polimerase em Tempo Real , Trypanosoma cruzi/genética , Trypanosoma cruzi/isolamento & purificação , Doenças Transmitidas por Vetores/diagnóstico , Doenças Transmitidas por Vetores/prevenção & controle , Doenças Transmitidas por Vetores/transmissão , Xenodiagnóstico
9.
J Med Entomol ; 59(1): 394-399, 2022 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-34448006

RESUMO

Chagas disease is caused by the infection of the parasite Trypanosoma cruzi (Chagas, 1909). Mexico is estimated to be among the countries with the highest rates of human infections. The southernmost region of the Baja California peninsula is home to the endemic, highly aggressive, and largest Triatominae vector, thus far described: Dipetalogaster maxima (Uhler 1894). Previous single-year studies have attempted to estimate the natural infection rate of T. cruzi in this species, none encompassing a multiyear sampling design nor a species-specific diagnostic tool. We report the infection rate based on more than 717 individuals examined via a PCR species-specific diagnosis. The infection rate of T. cruzi was of 4.4% (n = 5/112), 0.9% (n = 4/411), and 4.6% (n = 9/194) for 2016, 2017, and 2018, respectively, resulting in an infection rate of 2% across all sites and years (n = 18/717).


Assuntos
Triatoma/parasitologia , Trypanosoma cruzi/isolamento & purificação , Animais , Doença de Chagas/transmissão , Humanos , Insetos Vetores/parasitologia , México , Prevalência , Reduviidae/parasitologia , Doenças Transmitidas por Vetores/transmissão
10.
PLoS Comput Biol ; 17(11): e1009467, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34797822

RESUMO

We present artificial neural networks as a feasible replacement for a mechanistic model of mosquito abundance. We develop a feed-forward neural network, a long short-term memory recurrent neural network, and a gated recurrent unit network. We evaluate the networks in their ability to replicate the spatiotemporal features of mosquito populations predicted by the mechanistic model, and discuss how augmenting the training data with time series that emphasize specific dynamical behaviors affects model performance. We conclude with an outlook on how such equation-free models may facilitate vector control or the estimation of disease risk at arbitrary spatial scales.


Assuntos
Aedes , Modelos Biológicos , Mosquitos Vetores , Redes Neurais de Computação , Aedes/virologia , Animais , Biologia Computacional , Bases de Dados Factuais/estatística & dados numéricos , Humanos , Mosquitos Vetores/virologia , Dinâmica Populacional/estatística & dados numéricos , Análise Espaço-Temporal , Processos Estocásticos , Análise de Sistemas , Estados Unidos/epidemiologia , Doenças Transmitidas por Vetores/epidemiologia , Doenças Transmitidas por Vetores/transmissão , Doenças Transmitidas por Vetores/virologia , Tempo (Meteorologia)
11.
Nat Commun ; 12(1): 6825, 2021 11 24.
Artigo em Inglês | MEDLINE | ID: mdl-34819517

RESUMO

Mosquito-borne diseases present a worldwide public health burden. Current efforts to understand and counteract them have been aided by the use of cultured mosquito cells. Moreover, application in mammalian cells of forward genetic approaches such as CRISPR screens have identified essential genes and genes required for host-pathogen interactions, and in general, aided in functional annotation of genes. An equivalent approach for genetic screening of mosquito cell lines has been lacking. To develop such an approach, we design a new bioinformatic portal for sgRNA library design in several mosquito genomes, engineer mosquito cell lines to express Cas9 and accept sgRNA at scale, and identify optimal promoters for sgRNA expression in several mosquito species. We then optimize a recombination-mediated cassette exchange system to deliver CRISPR sgRNA and perform pooled CRISPR screens in an Anopheles cell line. Altogether, we provide a platform for high-throughput genome-scale screening in cell lines from disease vector species.


Assuntos
Sistemas CRISPR-Cas/genética , Controle de Mosquitos/métodos , Mosquitos Vetores/genética , Controle Biológico de Vetores/métodos , Doenças Transmitidas por Vetores/prevenção & controle , Animais , Anopheles/genética , Linhagem Celular , Biologia Computacional/métodos , Técnicas de Inativação de Genes , Biblioteca Gênica , Genes Essenciais , Humanos , RNA Guia de Cinetoplastídeos/genética , Doenças Transmitidas por Vetores/transmissão
13.
Viruses ; 13(11)2021 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-34835050

RESUMO

Following the recent outbreak of Zika virus (ZIKV) infections in Latin America, ZIKV has emerged as a global health threat due to its ability to induce neurological disease in both adults and the developing fetus. ZIKV is largely mosquito-borne and is now endemic in many parts of Africa, Asia, and South America. However, several reports have demonstrated persistent ZIKV infection of the male reproductive tract and evidence of male-to-female sexual transmission of ZIKV. Sexual transmission may broaden the reach of ZIKV infections beyond its current geographical limits, presenting a significant threat worldwide. Several mouse models of ZIKV infection have been developed to investigate ZIKV pathogenesis and develop effective vaccines and therapeutics. However, the majority of these models focus on mosquito-borne infection, while few have considered the impact of sexual transmission on immunity and pathogenesis. This review will examine the advantages and disadvantages of current models of mosquito-borne and sexually transmitted ZIKV and provide recommendations for the effective use of ZIKV mouse models.


Assuntos
Modelos Animais de Doenças , Doenças Virais Sexualmente Transmissíveis/transmissão , Infecção por Zika virus/transmissão , Animais , Feminino , Camundongos , Gravidez , Complicações Infecciosas na Gravidez/prevenção & controle , Complicações Infecciosas na Gravidez/virologia , Doenças Virais Sexualmente Transmissíveis/prevenção & controle , Doenças Transmitidas por Vetores/prevenção & controle , Doenças Transmitidas por Vetores/transmissão , Doenças Transmitidas por Vetores/virologia , Zika virus/fisiologia , Infecção por Zika virus/prevenção & controle , Infecção por Zika virus/virologia
14.
Parasit Vectors ; 14(1): 527, 2021 Oct 11.
Artigo em Inglês | MEDLINE | ID: mdl-34635176

RESUMO

The expansion of mosquito-borne diseases such as dengue, yellow fever, and chikungunya in the past 15 years has ignited the need for active surveillance of common and neglected mosquito-borne infectious diseases. The surveillance should be designed to detect diseases and to provide relevant field-based data for developing and implementing effective control measures to prevent outbreaks before significant public health consequences can occur. Mosquitoes are important vectors of human and animal pathogens, and knowledge on their biodiversity and distribution in the Afrotropical region is needed for the development of evidence-based vector control strategies. Following a comprehensive literature search, an inventory of the diversity and distribution of mosquitoes as well as the different mosquito-borne diseases found in Cameroon was made. A total of 290 publications/reports and the mosquito catalogue website were consulted for the review. To date, about 307 species, four subspecies and one putative new species of Culicidae, comprising 60 species and one putative new species of Anopheles, 67 species and two subspecies of Culex, 77 species and one subspecies of Aedes, 31 species and one subspecies of Eretmapodites, two Mansonia, eight Coquillettidia, and 62 species with unknown medical and veterinary importance (Toxorhynchites, Uranotaenia, Mimomyia, Malaya, Hodgesia, Ficalbia, Orthopodomyia, Aedeomyia, and Culiseta and Lutzia) have been collected in Cameroon. Multiple mosquito species implicated in the transmission of pathogens within Anopheles, Culex, Aedes, Eretmapodites, Mansonia, and Coquillettidia have been reported in Cameroon. Furthermore, the presence of 26 human and zoonotic arboviral diseases, one helminthic disease, and two protozoal diseases has been reported. Information on the bionomics, taxonomy, and distribution of mosquito species will be useful for the development of integrated vector management programmes for the surveillance and elimination of mosquito-borne diseases in Cameroon.


Assuntos
Culicidae/fisiologia , Mosquitos Vetores/fisiologia , Doenças Transmitidas por Vetores/transmissão , Aedes/parasitologia , Aedes/fisiologia , Aedes/virologia , Animais , Anopheles/parasitologia , Anopheles/fisiologia , Anopheles/virologia , Camarões , Culex/parasitologia , Culex/fisiologia , Culex/virologia , Culicidae/classificação , Culicidae/parasitologia , Culicidae/virologia , Surtos de Doenças , Humanos , Mosquitos Vetores/classificação , Doenças Transmitidas por Vetores/parasitologia , Doenças Transmitidas por Vetores/virologia
15.
Proc Natl Acad Sci U S A ; 118(41)2021 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-34620712

RESUMO

Wolbachia bacteria, inherited through the female germ line, infect a large fraction of arthropod species. Many Wolbachia strains manipulate host reproduction, most commonly through cytoplasmic incompatibility (CI). CI, a conditional male sterility, results when Wolbachia-infected male insects mate with uninfected females; viability is restored if the female is similarly infected (called "rescue"). CI is used to help control mosquito-borne viruses such as dengue and Zika, but its mechanisms remain unknown. The coexpressed CI factors CifA and CifB form stable complexes in vitro, but the timing and function of this interaction in the insect are unresolved. CifA expression in the female germ line is sufficient for rescue. We report high-resolution structures of a CI-factor complex, CinA-CinB, which utilizes a unique binding mode between the CinA rescue factor and the CinB nuclease; the structures were validated by biochemical and yeast growth analyses. Importantly, transgenic expression in Drosophila of a nonbinding CinA mutant, designed based on the CinA-CinB structure, suggests CinA expressed in females must bind CinB imported by sperm in order to rescue embryonic viability. Binding between cognate factors is conserved in an enzymatically distinct CI system, CidA-CidB, suggesting universal features in Wolbachia CI induction and rescue.


Assuntos
Drosophila melanogaster/microbiologia , Embrião não Mamífero/embriologia , Infertilidade Masculina/fisiopatologia , Reprodução/fisiologia , Wolbachia/metabolismo , Animais , Animais Geneticamente Modificados , Drosophila melanogaster/genética , Desenvolvimento Embrionário , Feminino , Masculino , Controle de Mosquitos/métodos , Complexos Multiproteicos/metabolismo , Ligação Proteica , Simbiose , Doenças Transmitidas por Vetores/prevenção & controle , Doenças Transmitidas por Vetores/transmissão , Doenças Transmitidas por Vetores/virologia
16.
Parasit Vectors ; 14(1): 509, 2021 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-34593023

RESUMO

BACKGROUND: Identifying the mechanisms driving disease risk is challenging for multi-host pathogens, such as Borrelia burgdorferi sensu lato (s.l.), the tick-borne bacteria causing Lyme disease. Deer are tick reproduction hosts but do not transmit B. burgdorferi s.l., whereas rodents and birds are competent transmission hosts. Here, we use a long-term deer exclosure experiment to test three mechanisms for how high deer density might shape B. burgdorferi s.l. prevalence in ticks: increased prevalence due to higher larval tick densities facilitating high transmission on rodents (M1); alternatively, reduced B. burgdorferi s.l. prevalence because more larval ticks feed on deer rather than transmission-competent rodents (dilution effect) (M2), potentially due to ecological cascades, whereby higher deer grazing pressure shortens vegetation which decreases rodent abundance thus reducing transmission (M3). METHODS: In a large enclosure where red deer stags were kept at high density (35.5 deer km-2), we used an experimental design consisting of eight plots of 0.23 ha, four of which were fenced to simulate the absence of deer and four that were accessible to deer. In each plot we measured the density of questing nymphs and nymphal infection prevalence in spring, summer and autumn, and quantified vegetation height and density, and small mammal abundance. RESULTS: Prevalence tended to be lower, though not conclusively so, in high deer density plots compared to exclosures (predicted prevalence of 1.0% vs 2.2%), suggesting that the dilution and cascade mechanisms might outweigh the increased opportunities for transmission mechanism. Presence of deer at high density led to shorter vegetation and fewer rodents, consistent with an ecological cascade. However, Lyme disease hazard (density of infected I. ricinus nymphs) was five times higher in high deer density plots due to tick density being 18 times higher. CONCLUSIONS: High densities of tick reproduction hosts such as deer can drive up vector-borne disease hazard, despite the potential to simultaneously reduce pathogen prevalence. This has implications for environmental pathogen management and for deer management, although the impact of intermediate deer densities now needs testing.


Assuntos
Borrelia burgdorferi/genética , Cervos/parasitologia , Ixodes/microbiologia , Doença de Lyme/epidemiologia , Doença de Lyme/transmissão , Infestações por Carrapato/veterinária , Distribuição Animal , Animais , Borrelia burgdorferi/fisiologia , Feminino , Larva/genética , Larva/microbiologia , Masculino , Prevalência , Roedores/parasitologia , Escócia/epidemiologia , Doenças Transmitidas por Vetores/epidemiologia , Doenças Transmitidas por Vetores/parasitologia , Doenças Transmitidas por Vetores/transmissão
17.
Comput Math Methods Med ; 2021: 2536720, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34646332

RESUMO

The aim of this work is to introduce a stochastic solver based on the Levenberg-Marquardt backpropagation neural networks (LMBNNs) for the nonlinear host-vector-predator model. The nonlinear host-vector-predator model is dependent upon five classes, susceptible/infected populations of host plant, susceptible/infected vectors population, and population of predator. The numerical performances through the LMBNN solver are observed for three different types of the nonlinear host-vector-predator model using the authentication, testing, sample data, and training. The proportions of these data are chosen as a larger part, i.e., 80% for training and 10% for validation and testing, respectively. The nonlinear host-vector-predator model is numerically treated through the LMBNNs, and comparative investigations have been performed using the reference solutions. The obtained results of the model are presented using the LMBNNs to reduce the mean square error (MSE). For the competence, exactness, consistency, and efficacy of the LMBNNs, the numerical results using the proportional measures through the MSE, error histograms (EHs), and regression/correlation are performed.


Assuntos
Modelos Biológicos , Redes Neurais de Computação , Doenças das Plantas/microbiologia , Animais , Biologia Computacional , Simulação por Computador , Vetores de Doenças , Dinâmica não Linear , Doenças das Plantas/estatística & dados numéricos , Comportamento Predatório , Processos Estocásticos , Doenças Transmitidas por Vetores/microbiologia , Doenças Transmitidas por Vetores/transmissão
18.
Parasit Vectors ; 14(1): 500, 2021 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-34565464

RESUMO

BACKGROUND: Vector-borne diseases (VBDs) such as dengue, chikungunya, and Zika pose a significant challenge to health systems in countries they affect, especially countries with less developed healthcare systems. Therefore, countries are encouraged to work towards more resilient health systems. This qualitative study aims to examine the performance of the health system of the Dutch Caribbean island of CuraÒ«ao regarding the prevention and control of VBDs in the last decade by using the WHO health system building blocks. METHODS: From November 2018 to December 2020, a multi-method qualitative study was performed in Curaçao, applying content analysis of documents (n = 50), five focus group discussions (n = 30), interviews with experts (n = 11) and 15 observation sessions. The study was designed based on the WHO framework: health system building blocks. Two cycles of inductive and deductive coding were employed, and Nvivo software was used to analyse the data. RESULTS: This study's data highlighted the challenges (e.g. insufficient oversight, coordination, leadership skills, structure and communication) that the departments of the health system of CuraÒ«ao faced during the last three epidemics of VBDs (2010-2020). Furthermore, low levels of collaboration between governmental and non-governmental organisations (e.g. semi-governmental and private laboratories) and insufficient capacity building to improve skills (e.g. entomological, surveillance skills) were also observed. Lastly, we observed how bottlenecks in one building block negatively influenced other building blocks (e.g. inadequate leadership/governance obstructed the workforce's performance). CONCLUSIONS: This study uncovers potential organisational bottlenecks that have affected the performance of the health system of CuraÒ«ao negatively. We recommend starting with the reinforcement of oversight of the integrated vector management programme to ensure the development, implementation and evaluation of related legislation, policies and interventions. Also, we recommend evaluating and reforming the existing administrative and organisational structure of the health system by considering the cultural style, challenges and barriers of the current health system. More efforts are needed to improve the documentation of agreements, recruitment and evaluation of the workforce's performance. Based on our findings, we conceptualised actions to strengthen the health system's building blocks to improve its performance for future outbreaks of infectious diseases.


Assuntos
Atenção à Saúde/normas , Doenças Transmitidas por Vetores/epidemiologia , Animais , Curaçao , Atenção à Saúde/organização & administração , Entomologia/métodos , Entomologia/normas , Mão de Obra em Saúde/normas , Humanos , Colaboração Intersetorial , Liderança , Mosquitos Vetores/fisiologia , Pesquisa Qualitativa , Doenças Transmitidas por Vetores/transmissão
19.
Viruses ; 13(6)2021 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-34208620

RESUMO

We describe the impact of COVID-19 mitigation measures on mosquito-borne diseases in Queensland, Australia, during the first half of 2020. Implementation of restrictions coincided with an atypical late season outbreak of Ross River virus (RRV) characterized by a peak in notifications in April (1173) and May (955) which were greater than 3-fold the mean observed for the previous four years. We propose that limitations on human movement likely resulted in the majority of RRV infections being acquired at or near the place of residence, and that an increase in outdoor activities, such as gardening and bushwalking in the local household vicinity, increased risk of exposure to RRV-infected mosquitoes. In contrast, the precipitous decline in international passenger flights led to a reduction in the number of imported dengue and malaria cases of over 70% and 60%, respectively, compared with the previous five years. This substantial reduction in flights also reduced a risk pathway for importation of exotic mosquitoes, but the risk posed by importation via sea cargo was not affected. Overall, the emergence of COVID-19 has had a varied impact on mosquito-borne disease epidemiology in Queensland, but the need for mosquito surveillance and control, together with encouragement of personal protective measures, remains unchanged.


Assuntos
COVID-19/prevenção & controle , Surtos de Doenças/prevenção & controle , Vigilância da População , Doenças Transmitidas por Vetores/epidemiologia , Infecções por Alphavirus/epidemiologia , Infecções por Alphavirus/transmissão , Animais , COVID-19/epidemiologia , Controle de Doenças Transmissíveis/métodos , Controle de Doenças Transmissíveis/estatística & dados numéricos , Culicidae/virologia , Surtos de Doenças/estatística & dados numéricos , Humanos , Movimento , Queensland/epidemiologia , Viagem , Doenças Transmitidas por Vetores/prevenção & controle , Doenças Transmitidas por Vetores/transmissão
20.
J Gen Virol ; 102(6)2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-34166178

RESUMO

Mosquito-transmitted arboviruses constitute a large proportion of emerging infectious diseases that are both a public health problem and a threat to animal populations. Many such viruses were identified in East Africa, a region where they remain important and from where new arboviruses may emerge. We set out to describe and review the relevant mosquito-borne viruses that have been identified specifically in Uganda. We focused on the discovery, burden, mode of transmission, animal hosts and clinical manifestation of those previously involved in disease outbreaks. A search for mosquito-borne arboviruses detected in Uganda was conducted using search terms 'Arboviruses in Uganda' and 'Mosquitoes and Viruses in Uganda' in PubMed and Google Scholar in 2020. Twenty-four mosquito-borne viruses from different animal hosts, humans and mosquitoes were documented. The majority of these were from family Peribunyaviridae, followed by Flaviviridae, Togaviridae, Phenuiviridae and only one each from family Rhabdoviridae and Reoviridae. Sixteen (66.7 %) of the viruses were associated with febrile illnesses. Ten (41.7 %) of them were first described locally in Uganda. Six of these are a public threat as they have been previously associated with disease outbreaks either within or outside Uganda. Historically, there is a high burden and endemicity of arboviruses in Uganda. Given the many diverse mosquito species known in the country, there is also a likelihood of many undescribed mosquito-borne viruses. New generation diagnostic platforms have great potential to identify new viruses. Indeed, four novel viruses, two of which were from humans (Ntwetwe and Nyangole viruses) and two from mosquitoes (Kibale and Mburo viruses) including the 2010 yellow fever virus (YFV) outbreak were identified in the last decade using next generation sequencing. Given the unbiased approach of detection of viruses by this technology, its use will undoubtedly be critically important in the characterization of mosquito viromes which in turn will inform other diagnostic efforts.


Assuntos
Infecções por Arbovirus , Arbovírus , Doenças Transmissíveis Emergentes/virologia , Mosquitos Vetores/virologia , Doenças Transmitidas por Vetores/virologia , Animais , Infecções por Arbovirus/epidemiologia , Infecções por Arbovirus/transmissão , Infecções por Arbovirus/veterinária , Infecções por Arbovirus/virologia , Arbovírus/classificação , Arbovírus/genética , Arbovírus/isolamento & purificação , Doenças Transmissíveis Emergentes/epidemiologia , Doenças Transmissíveis Emergentes/transmissão , Doenças Transmissíveis Emergentes/veterinária , Culicidae/virologia , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Uganda/epidemiologia , Doenças Transmitidas por Vetores/epidemiologia , Doenças Transmitidas por Vetores/transmissão , Doenças Transmitidas por Vetores/veterinária
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...